Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Josefa Chuh is active.

Publication


Featured researches published by Josefa Chuh.


Bioconjugate Chemistry | 2011

Impact of drug conjugation on pharmacokinetics and tissue distribution of anti-STEAP1 antibody-drug conjugates in rats.

C. Andrew Boswell; Eduardo E. Mundo; Crystal Zhang; Daniela Bumbaca; Nicole R. Valle; Katherine R. Kozak; Aimee Fourie; Josefa Chuh; Neelima Koppada; Ola Saad; Herman S. Gill; Ben-Quan Shen; Bonnee Rubinfeld; Jay Tibbitts; Surinder Kaur; Frank-Peter Theil; Paul J. Fielder; Leslie A. Khawli; Kedan Lin

Antibody-drug conjugates (ADCs) are designed to combine the exquisite specificity of antibodies to target tumor antigens with the cytotoxic potency of chemotherapeutic drugs. In addition to the general chemical stability of the linker, a thorough understanding of the relationship between ADC composition and biological disposition is necessary to ensure that the therapeutic window is not compromised by altered pharmacokinetics (PK), tissue distribution, and/or potential organ toxicity. The six-transmembrane epithelial antigen of prostate 1 (STEAP1) is being pursued as a tumor antigen target. To assess the role of ADC composition in PK, we evaluated plasma and tissue PK profiles in rats, following a single dose, of a humanized anti-STEAP1 IgG1 antibody, a thio-anti-STEAP1 (ThioMab) variant, and two corresponding thioether-linked monomethylauristatin E (MMAE) drug conjugates modified through interchain disulfide cysteine residues (ADC) and engineered cysteines (TDC), respectively. Plasma PK of total antibody measured by enzyme-linked immunosorbent assay (ELISA) revealed ∼45% faster clearance for the ADC relative to the parent antibody, but no apparent difference in clearance between the TDC and unconjugated parent ThioMab. Total antibody clearances of the two unconjugated antibodies were similar, suggesting minimal effects on PK from cysteine mutation. An ELISA specific for MMAE-conjugated antibody indicated that the ADC cleared more rapidly than the TDC, but total antibody ELISA showed comparable clearance for the two drug conjugates. Furthermore, consistent with relative drug load, the ADC had a greater magnitude of drug deconjugation than the TDC in terms of free plasma MMAE levels. Antibody conjugation had a noticeable, albeit minor, impact on tissue distribution with a general trend toward increased hepatic uptake and reduced levels in other highly vascularized organs. Liver uptakes of ADC and TDC at 5 days postinjection were 2-fold and 1.3-fold higher, respectively, relative to the unmodified antibodies. Taken together, these results indicate that the degree of overall structural modification in anti-STEAP1-MMAE conjugates has a corresponding level of impact on both PK and tissue distribution.


Molecular Cancer Therapeutics | 2012

Effects of Anti-VEGF on Pharmacokinetics, Biodistribution, and Tumor Penetration of Trastuzumab in a Preclinical Breast Cancer Model

Cinthia V. Pastuskovas; Eduardo E. Mundo; Simon Williams; Tapan K Nayak; Jason Ho; Sheila Ulufatu; Suzanna Clark; Sarajane Ross; Eric Cheng; Kathryn Parsons-Reponte; Gary Cain; Marjie Van Hoy; Nicholas Majidy; Sheila Bheddah; Josefa Chuh; Katherine R. Kozak; Nicholas Lewin-Koh; Peter Nauka; Daniela Bumbaca; Mark X. Sliwkowski; Jay Tibbitts; Frank-Peter Theil; Paul J. Fielder; Leslie A. Khawli; C. Andrew Boswell

Both human epidermal growth factor receptor 2 (HER-2/neu) and VEGF overexpression correlate with aggressive phenotypes and decreased survival among breast cancer patients. Concordantly, the combination of trastuzumab (anti-HER2) with bevacizumab (anti-VEGF) has shown promising results in preclinical xenograft studies and in clinical trials. However, despite the known antiangiogenic mechanism of anti-VEGF antibodies, relatively little is known about their effects on the pharmacokinetics and tissue distribution of other antibodies. This study aimed to measure the disposition properties, with a particular emphasis on tumor uptake, of trastuzumab in the presence or absence of anti-VEGF. Radiolabeled trastuzumab was administered alone or in combination with an anti-VEGF antibody to mice bearing HER2-expressing KPL-4 breast cancer xenografts. Biodistribution, autoradiography, and single-photon emission computed tomography–X-ray computed tomography imaging all showed that anti-VEGF administration reduced accumulation of trastuzumab in tumors despite comparable blood exposures and similar distributions in most other tissues. A similar trend was also observed for an isotype-matched IgG with no affinity for HER2, showing reduced vascular permeability to macromolecules. Reduced tumor blood flow (P < 0.05) was observed following anti-VEGF treatment, with no significant differences in the other physiologic parameters measured despite immunohistochemical evidence of reduced vascular density. In conclusion, anti-VEGF preadministration decreased tumor uptake of trastuzumab, and this phenomenon was mechanistically attributed to reduced vascular permeability and blood perfusion. These findings may ultimately help inform dosing strategies to achieve improved clinical outcomes. Mol Cancer Ther; 11(3); 752–62. ©2012 AACR.


Journal of Medicinal Chemistry | 2014

Site-Specific Trastuzumab Maytansinoid Antibody–Drug Conjugates with Improved Therapeutic Activity through Linker and Antibody Engineering

Thomas H. Pillow; Janet Tien; Kathryn Parsons-Reponte; Sunil Bhakta; Hao Li; Leanna Staben; Guangmin Li; Josefa Chuh; Aimee Fourie-O’Donohue; Martine Darwish; Victor Yip; Luna Liu; Douglas D. Leipold; Dian Su; Elmer Wu; Susan D. Spencer; Ben-Quan Shen; Keyang Xu; Katherine R. Kozak; Helga Raab; Richard Vandlen; Gail Lewis Phillips; Richard H. Scheller; Paul Polakis; Mark X. Sliwkowski; John A. Flygare; Jagath R. Junutula

Antibody-drug conjugates (ADCs) have a significant impact toward the treatment of cancer, as evidenced by the clinical activity of the recently approved ADCs, brentuximab vedotin for Hodgkin lymphoma and ado-trastuzumab emtansine (trastuzumab-MCC-DM1) for metastatic HER2+ breast cancer. DM1 is an analog of the natural product maytansine, a microtubule inhibitor that by itself has limited clinical activity and high systemic toxicity. However, by conjugation of DM1 to trastuzumab, the safety was improved and clinical activity was demonstrated. Here, we report that through chemical modification of the linker-drug and antibody engineering, the therapeutic activity of trastuzumab maytansinoid ADCs can be further improved. These improvements include eliminating DM1 release in the plasma and increasing the drug load by engineering four cysteine residues into the antibody. The chemical synthesis of highly stable linker-drugs and the modification of cysteine residues of engineered site-specific antibodies resulted in a homogeneous ADC with increased therapeutic activity compared to the clinically approved ADC, trastuzumab-MCC-DM1.


Molecular Cancer Therapeutics | 2013

DCDT2980S, an Anti-CD22-Monomethyl Auristatin E Antibody–Drug Conjugate, Is a Potential Treatment for Non-Hodgkin Lymphoma

Dongwei Li; Kirsten Achilles Poon; Shang-Fan Yu; Randall Dere; MaryAnn Go; Jeffrey Lau; Bing Zheng; Kristi Elkins; Dimitry M. Danilenko; Katherine R. Kozak; Pamela Chan; Josefa Chuh; Xiaoyan Shi; Denise Nazzal; Franklin Fuh; Jacqueline McBride; Vanitha Ramakrishnan; Ruth de Tute; Andy C. Rawstron; Andrew Jack; Rong Deng; Yu-Waye Chu; David Dornan; Marna Williams; William Ho; Allen Ebens; Saileta Prabhu; Andrew G. Polson

Antibody–drug conjugates (ADC), potent cytotoxic drugs linked to antibodies via chemical linkers, allow specific targeting of drugs to neoplastic cells. We have used this technology to develop the ADC DCDT2980S that targets CD22, an antigen with expression limited to B cells and the vast majority of non-Hodgkin lymphomas (NHL). DCDT2980S consists of a humanized anti-CD22 monoclonal IgG1 antibody with a potent microtubule-disrupting agent, monomethyl auristatin E (MMAE), linked to the reduced cysteines of the antibody via a protease cleavable linker, maleimidocaproyl-valine-citrulline-p-aminobenzoyloxycarbonyl (MC-vc-PAB). We describe the efficacy, safety, and pharmacokinetics of DCDT2980S in animal models to assess its potential as a therapeutic for the treatment of B-cell malignancies. We did not find a strong correlation between in vitro or in vivo efficacy and CD22 surface expression, nor a correlation of sensitivity to free drug and in vitro potency. We show that DCDT2980S was capable of inducing complete tumor regression in xenograft mouse models of NHL and can be more effective than rituximab plus combination chemotherapy at drug exposures that were well tolerated in cynomolgus monkeys. These results suggest that DCDT2980S has an efficacy, safety, and pharmacokinetics profile that support potential treatment of NHL. Mol Cancer Ther; 12(7); 1255–65. ©2013 AACR.


Nature Chemistry | 2016

Targeted drug delivery through the traceless release of tertiary and heteroaryl amines from antibody–drug conjugates

Leanna Staben; Stefan G. Koenig; Sophie M. Lehar; Richard Vandlen; Donglu Zhang; Josefa Chuh; Shang-Fan Yu; Carl Ng; Jun Guo; Yanzhou Liu; Aimee Fourie-O'Donohue; MaryAnn Go; Xin Linghu; Nathaniel L. Segraves; Tao Wang; Jinhua Chen; Binqing Wei; Gail Lewis Phillips; Keyang Xu; Katherine R. Kozak; Sanjeev Mariathasan; John A. Flygare; Thomas H. Pillow

The reversible attachment of a small-molecule drug to a carrier for targeted delivery can improve pharmacokinetics and the therapeutic index. Previous studies have reported the delivery of molecules that contain primary and secondary amines via an amide or carbamate bond; however, the ability to employ tertiary-amine-containing bioactive molecules has been elusive. Here we describe a bioreversible linkage based on a quaternary ammonium that can be used to connect a broad array of tertiary and heteroaryl amines to a carrier protein. Using a concise, protecting-group-free synthesis we demonstrate the chemoselective modification of 12 complex molecules that contain a range of reactive functional groups. We also show the utility of this connection with both protease-cleavable and reductively cleavable antibody-drug conjugates that were effective and stable in vitro and in vivo. Studies with a tertiary-amine-containing antibiotic show that the resulting antibody-antibiotic conjugate provided appropriate stability and release characteristics and led to an unexpected improvement in activity over the conjugates previously connected via a carbamate.


Molecular Cancer Therapeutics | 2014

An Antimesothelin-Monomethyl Auristatin E Conjugate with Potent Antitumor Activity in Ovarian, Pancreatic, and Mesothelioma Models

Suzie J. Scales; Nidhi Gupta; Glenn Pacheco; Ron Firestein; Dorothy French; Hartmut Koeppen; Linda Rangell; Vivian Barry-Hamilton; Elizabeth Luis; Josefa Chuh; Yin Zhang; Gladys Ingle; Aimee Fourie-O'Donohue; Katherine R. Kozak; Sarajane Ross; Mark S. Dennis; Susan D. Spencer

Mesothelin (MSLN) is an attractive target for antibody–drug conjugate therapy because it is highly expressed in various epithelial cancers, with normal expression limited to nondividing mesothelia. We generated novel antimesothelin antibodies and conjugated an internalizing one (7D9) to the microtubule-disrupting drugs monomethyl auristatin E (MMAE) and MMAF, finding the most effective to be MMAE with a lysosomal protease-cleavable valine–citrulline linker. The humanized (h7D9.v3) version, αMSLN-MMAE, specifically targeted mesothelin-expressing cells and inhibited their proliferation with an IC50 of 0.3 nmol/L. Because the antitumor activity of an antimesothelin immunotoxin (SS1P) in transfected mesothelin models did not translate to the clinic, we carefully selected in vivo efficacy models endogenously expressing clinically relevant levels of mesothelin, after scoring mesothelin levels in ovarian, pancreatic, and mesothelioma tumors by immunohistochemistry. We found that endogenous mesothelin in cancer cells is upregulated in vivo and identified two suitable xenograft models for each of these three indications. A single dose of αMSLN-MMAE profoundly inhibited or regressed tumor growth in a dose-dependent manner in all six models, including two patient-derived tumor xenografts. The robust and durable efficacy of αMSLN-MMAE in preclinical models of ovarian, mesothelioma, and pancreatic cancers justifies the ongoing phase I clinical trial. Mol Cancer Ther; 13(11); 2630–40. ©2014 AACR.


Cancer Research | 2017

Cathepsin B Is Dispensable for Cellular Processing of Cathepsin B-Cleavable Antibody–Drug Conjugates

Niña G. Caculitan; Josefa Chuh; Yong Ma; Donglu Zhang; Katherine R. Kozak; Yichin Liu; Thomas H. Pillow; Jack Sadowsky; Tommy K. Cheung; Qui Phung; Benjamin Haley; Byoung-Chul Lee; Robert W. Akita; Mark X. Sliwkowski; Andrew G. Polson

Antibody-drug conjugates (ADC) are designed to selectively bind to tumor antigens via the antibody and release their cytotoxic payload upon internalization. Controllable payload release through judicious design of the linker has been an early technological milestone. Here, we examine the effect of the protease-cleavable valine-citrulline [VC(S)] linker on ADC efficacy. The VC(S) linker was designed to be cleaved by cathepsin B, a lysosomal cysteine protease. Surprisingly, suppression of cathepsin B expression via CRISPR-Cas9 gene deletion or shRNA knockdown had no effect on the efficacy of ADCs with VC(S) linkers armed with a monomethyl auristatin E (MMAE) payload. Mass spectrometry studies of payload release suggested that other cysteine cathepsins can cleave the VC(S) linker. Also, ADCs with a nonprotease-cleavable enantiomer, the VC(R) isomer, mediated effective cell killing with a cysteine-VC(R)-MMAE catabolite generated by lysosomal catabolism. Based on these observations, we altered the payload to a pyrrolo[2,1-c][1,4]benzodiazepine dimer (PBD) conjugate that requires linker cleavage in order to bind its DNA target. Unlike the VC-MMAE ADCs, the VC(S)-PBD ADC is at least 20-fold more cytotoxic than the VC(R)-PBD ADC. Our findings reveal that the VC(S) linker has multiple paths to produce active catabolites and that antibody and intracellular targets are more critical to ADC efficacy. These results suggest that protease-cleavable linkers are unlikely to increase the therapeutic index of ADCs and that resistance based on linker processing is improbable. Cancer Res; 77(24); 7027-37. ©2017 AACR.


Journal of Medicinal Chemistry | 2017

Discovery of Peptidomimetic Antibody–Drug Conjugate Linkers with Enhanced Protease Specificity

Binqing Wei; Janet Gunzner-Toste; Hui Yao; Tao Wang; Jing Wang; Zijin Xu; Jinhua Chen; John S. Wai; Jim Nonomiya; Siao Ping Tsai; Josefa Chuh; Katherine R. Kozak; Yichin Liu; Shang-Fan Yu; Jeff Lau; Guangmin Li; Gail D. Phillips; Doug Leipold; Amrita Kamath; Dian Su; Keyang Xu; Charles Eigenbrot; Stefan Steinbacher; Rachana Ohri; Helga Raab; Leanna Staben; Guiling Zhao; John A. Flygare; Thomas H. Pillow; Vishal A. Verma

Antibody-drug conjugates (ADCs) have become an important therapeutic modality for oncology, with three approved by the FDA and over 60 others in clinical trials. Despite the progress, improvements in ADC therapeutic index are desired. Peptide-based ADC linkers that are cleaved by lysosomal proteases have shown sufficient stability in serum and effective payload-release in targeted cells. If the linker can be preferentially hydrolyzed by tumor-specific proteases, safety margin may improve. However, the use of peptide-based linkers limits our ability to modulate protease specificity. Here we report the structure-guided discovery of novel, nonpeptidic ADC linkers. We show that a cyclobutane-1,1-dicarboxamide-containing linker is hydrolyzed predominantly by cathepsin B while the valine-citrulline dipeptide linker is not. ADCs bearing the nonpeptidic linker are as efficacious and stable in vivo as those with the dipeptide linker. Our results strongly support the application of the peptidomimetic linker and present new opportunities for improving the selectivity of ADCs.


Cancer Research | 2014

Abstract 4502: Nonclinical characterization and tolerability of a surrogate anti-mesothelin-MMAE antibody-drug conjugate

Nidhi Gupta; Willy Solis; Reina N. Fuji; Amy Oldendorp; Glenn Pacheco; Elizabeth Luis; Josefa Chuh; Dorothy French; Elizabeth Drake; Mark S. Dennis; Katherine R. Kozak; Sarajane Ross; Jay Tibbitts; Susan D. Spencer; Suzie J. Scales

Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA Mesothelin (MSLN) is a cell surface glycoprotein widely expressed in several cancers with normal expression limited to the serosal mesothelia, features of an ideal target for antibody-based therapy. SS1P, a (dsFv)-PE38 immunotoxin to MSLN that kills cells by inhibition of protein synthesis, exhibits minor clinical responses as monotherapy, but was dose-limited by pleuritis, likely an antigen-dependent toxicity (Hassan et al., (2007) Clin Cancer Res 13 p3144). As mesothelial cells divide infrequently, we evaluated whether an anti-mitotic antibody-drug conjugate (ADC) directed to MSLN would be better tolerated. Our humanized lead anti-MSLN-MMAE (mc-vc-PAB-monomethylauristatin E) conjugate shows excellent preclinical activity (see accompanying abstract by Scales et al), but is specific to human MSLN, so a surrogate ADC that cross-reacts with cynomolgus monkey and rat MSLN was generated for non-clinical toxicity studies. While both the lead and the surrogate antibodies recognize human MSLN, they bound to different epitopes and the surrogate binding was sensitive to glycosylation, only recognizing a subset of cell lines expressing human MSLN with high affinity. The affinity of the surrogate antibody for cynomolgus monkey MSLN is 5 to 22-fold lower than that of the lead antibody for human MSLN, depending on its glycosylation pattern in the human parental cell line to which it was compared. Nonetheless, the surrogate antibody detects endogenous MSLN in monkey pleura and the surrogate ADC exhibits robust cytotoxic activity against monkey MSLN-expressing cells in vitro. Importantly, the surrogate ADC demonstrated comparable in vivo efficacy to the lead ADC against BJAB xenografts expressing monkey or human MSLN respectively, thus validating its use in safety studies. We thus conducted a repeat-dose monkey toxicity study with a clinically relevant (q3w x5) dosing schedule, which yielded similar results for both surrogate and lead ADCs (the primary finding being reversible myelotoxicity, an antigen-independent toxicity similar to those of other IgG1-MMAE ADCs (Li et al., (2013), Mol Can Thera 12 p1255)). Unlike SS1P, there was no evidence of target-dependent pleuritis, nor any other serositis. Our data suggest that anti-MSLN-MMAE ADCs may be safer than SS1P and helped define the Phase I starting dose. Citation Format: Nidhi Gupta, Willy A. Solis, Reina N. Fuji, Amy Oldendorp, Glenn Pacheco, Elizabeth Luis, Josefa Chuh, Dorothy M. French, Elizabeth Drake, Mark S. Dennis, Katherine R. Kozak, Sarajane Ross, Jay Tibbitts, Susan D. Spencer, Suzie J. Scales. Nonclinical characterization and tolerability of a surrogate anti-mesothelin-MMAE antibody-drug conjugate. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 4502. doi:10.1158/1538-7445.AM2014-4502


Cancer Research | 2014

Abstract 4494: A clinical candidate anti-mesothelin-MMAE antibody-drug conjugate (ADC) for therapy of mesothelin-expressing cancers

Suzie J. Scales; Nidhi Gupta; Glenn Pacheco; Ron Firestein; Dorothy French; Josefa Chuh; Yin Zhang; Leanne Berry; Jenny Bostrom; Elizabeth Luis; Aimee Fourie O'Donohue; Katherine R. Kozak; Sarajane Ross; Mark S. Dennis; Jay Tibbitts; Susan D. Spencer

Mesothelin (MSLN) is a cell surface glycoprotein widely expressed in a variety of cancers, with prevalence levels by IHC of 85% in ovarian, 75% in pancreatic and 45% in mesothelioma, and normal expression limited to the mesothelia, suggesting it could be an ideal target for antibody-drug conjugate therapy of these cancers. We have generated a high affinity (subnanomolar), humanized antibody to MSLN and conjugated it to auristatin anti-mitotic drugs (monomethylauristatin E and F, Seattle Genetics) via an uncleavable linker (anti-MSLN-mc-MMAF) or a cathepsin-cleavable valine-citrulline linker (anti-MSLN-mc-vc-PAB-MMAE/F) for comparison. The in vivo efficacy obtained in an ovarian transplant model was superior with the anti-MSLN-MMAE ADC. Anti-MSLN-MMAE was specifically internalized by MSLN-expressing cells in vitro, resulting in cell death compared to control ADCs. Pancreatic, ovarian and mesothelioma tumor cell lines endogenously expressing physiological levels of MSLN were identified and established as xenografts in mice. A single dose of anti-MSLN ADC was sufficient to inhibit or shrink tumor growth in models of each of the three indications in vivo, as well as inducing complete regressions in primary human pancreatic models, even those expressing low levels of MSLN typical of most human pancreatic tumors. Additionally, anti-MSLN-MMAE (at suboptimal doses) appeared to synergize with gemcitabine at clinically relevant doses in an HPAC xenograft model. Furthermore, anti-MSLN-MMAE was well tolerated in non-clinical toxicity studies (see accompanying abstract by Gupta et al.). Our data suggest that anti-MSLN-vc-MMAE is a promising clinical candidate for the treatment of several types of mesothelin-positive cancers. Citation Format: Suzie J. Scales, Nidhi Gupta, Glenn Pacheco, Ron Firestein, Dorothy M. French, Josefa Chuh, Yin Zhang, Leanne Berry, Jenny Bostrom, Elizabeth Luis, Aimee Fourie O9Donohue, Katherine R. Kozak, Sarajane Ross, Mark S. Dennis, Jay Tibbitts, Susan D. Spencer. A clinical candidate anti-mesothelin-MMAE antibody-drug conjugate (ADC) for therapy of mesothelin-expressing cancers. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 4494. doi:10.1158/1538-7445.AM2014-4494

Collaboration


Dive into the Josefa Chuh's collaboration.

Researchain Logo
Decentralizing Knowledge